1
|
Liu C, Zhang X, Xie Q, Fang B, Liu F, Luo J, Aihemaiti G, Ji W, Yang Y, Li X. Identification of key proteins and pathways in myocardial infarction using machine learning approaches. Sci Rep 2025; 15:19530. [PMID: 40467768 PMCID: PMC12137932 DOI: 10.1038/s41598-025-04401-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
Acute myocardial infarction (AMI) is a leading cause of global morbidity and mortality, requiring deeper insights into its molecular mechanisms for improved diagnosis and treatment. This study combines proteomics, transcriptomics and machine learning (ML) to identify key proteins and pathways associated with AMI. Plasma samples from 48 AMI patients and 50 healthy controls (HC) were used for proteomic sequencing. Differentially expressed proteins (DEPs) were identified and analyzed for pathway enrichment. Protein-protein interaction (PPI) networks were constructed, and we conducted a meta-analysis (GSE60993, GSE61144, GSE48060) using an inverse variance model to combine differentially expressed genes (DEGs) identified via LIMMA and FDR adjustment across three studies. Clustering and co-expression analysis were performed using K-Medoids and weighted gene co-expression network analysis (WGCNA). ML feature selection identified hub proteins, which were validated across bulk, single-cell, and spatial datasets for atherosclerosis (ATH) and MI. In this study, we identified 437 DEPs with 291 up-regulated and 146 down-regulated proteins. Functional enrichment analysis revealed key pathways involved in inflammation, immunity, metabolism, and cellular stress responses, among others. Using non-negative matrix factorization (NNMF) and K-Medoids clustering, AMI patients were divided into two clusters (C1 and C2), with distinct protein expression patterns and inflammatory responses. Differential analysis between clusters revealed 200 cluster-specific DEPs, with C1 associated with angiogenesis and vascular remodeling, and C2 linked to cellular stress and apoptosis. A meta-analysis identified 1383 DEGs, and their intersection with DEPs yielded 63 proteins, which were subsequently refined by logistic regression to 36 AMI-associated proteins. Furthermore, a protein co-expression network analysis identified 49 modules, with the turquoise module being strongly associated with AMI highlighting pathways in lipid metabolism, immune response, and tissue repair. From this module, 17 key proteins were selected, and ML further distilled these to nine core features (CAMP, CLTC, CTNNB1, FUBP3, IQGAP1, MANBA, ORM1, PSME1, and SPP1) that are closely linked to immune regulation, apoptosis, and metabolism. These proteins were validated across multiple datasets. Single-cell analysis revealed distinct expression patterns of these proteins across cell types and spatial regions in ATH and MI, emphasizing their roles in inflammation, vascular remodeling, and plaque instability. This study identifies critical proteins and pathways in AMI, offering potential biomarkers and therapeutic targets. The use of ML provides a robust framework for identifying AMI's key molecular.
Collapse
Affiliation(s)
- Chang Liu
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Xing Zhang
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Qian Xie
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Binbin Fang
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Fen Liu
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Junyi Luo
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Gulandanmu Aihemaiti
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Wei Ji
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Yining Yang
- Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, 830001, China.
| | - Xiaomei Li
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China.
| |
Collapse
|
2
|
Mesfin JM, Carrow KP, Chen A, Hopps MP, Holm JJ, Lyons QP, Nguyen MB, Hunter JD, Magassa A, Wong EG, Reimold K, Paleti SN, Gardner E, Thompson MP, Luo CG, Zhang X, Christman KL, Gianneschi NC. Protein-Like Polymers Targeting Keap1/Nrf2 as Therapeutics for Myocardial Infarction. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2417885. [PMID: 40277240 DOI: 10.1002/adma.202417885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/13/2025] [Indexed: 04/26/2025]
Abstract
Myocardial infarction (MI) results in oxidative stress to the myocardium and frequently leads to heart failure (HF). There is an unmet clinical need to develop therapeutics that address the inflammatory stress response and prevent negative left ventricular remodeling. Here, the Keap1/Nrf2 protein-protein interaction is specifically targeted, as Nrf2 activation is known to mitigate the inflammatory response following MI. This is achieved using a Nrf2-mimetic protein-like polymer (PLP) to inhibit the Keap1-Nrf2 interaction. The PLP platform technology provides stability in vivo, potent intracellular bioactivity, and multivalency leading to high avidity Keap1 binding. In vitro and in vivo assays to probe cellular activity and MI therapeutic utility are employed. These Keap1-inhibiting PLPs (Keap1i-PLPs) impart cytoprotection from oxidative stress via Nrf2 activation at sub-nanomolar concentrations in primary cardiomyocytes. Single-digit mg kg-1, single-dose, intravenous PLP administration significantly improves cardiac function in rats post-MI through immunomodulatory, anti-apoptotic, and angiogenic mechanisms. Thus Keap1i-PLPs disrupt key intracellular protein-protein interactions following intravenous, systemic administration in vivo. These results have broad implications not only for MI but also for other oxidative stress-driven diseases and conditions.
Collapse
Affiliation(s)
- Joshua M Mesfin
- Shu Chien-Gene Lay Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92037, USA
| | - Kendal P Carrow
- Medical Scientist Training Program, Department of Biomedical Engineering, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Alexander Chen
- Program in Materials Science and Engineering, Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92037, USA
| | - Madeline P Hopps
- Department of Chemistry, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - JoJo J Holm
- Department of Chemistry, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Quincy P Lyons
- Shu Chien-Gene Lay Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92037, USA
| | - Michael B Nguyen
- Shu Chien-Gene Lay Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92037, USA
| | - Jervaughn D Hunter
- Shu Chien-Gene Lay Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92037, USA
| | - Assa Magassa
- Department of Chemistry, Department of Materials Science & Engineering, Department of Pharmacology, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Elyse G Wong
- Shu Chien-Gene Lay Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92037, USA
| | - Kate Reimold
- Shu Chien-Gene Lay Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92037, USA
| | - Sriya N Paleti
- Shu Chien-Gene Lay Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92037, USA
| | - Emily Gardner
- Shu Chien-Gene Lay Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92037, USA
| | - Matthew P Thompson
- Department of Chemistry, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Colin G Luo
- Shu Chien-Gene Lay Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92037, USA
| | - Xiaoyu Zhang
- Department of Chemistry, Department of Materials Science & Engineering, Department of Pharmacology, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Karen L Christman
- Shu Chien-Gene Lay Department of Bioengineering, Program in Materials Science and Engineering, Sanford Consortium for Regenerative Medicine, Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA, 92037, USA
| | - Nathan C Gianneschi
- Department of Biomedical Engineering, Department of Chemistry, Department of Materials Science & Engineering, Department of Pharmacology, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
3
|
Kakaei Y, Hussain S, Elmahdy A, Berger E, Shekka Espinosa A, Sevastianova V, Sheybani Z, Al-Awar A, Kalani M, Jha S, Zulfaj E, Nejat A, Jha A, Pylova T, Krasnikova M, Andersson EA, Silva VRR, Omerovic E, Redfors B. Comparison of the proteomic landscape in experimental ischemia reperfusion with versus without ischemic preconditioning. Sci Rep 2025; 15:11836. [PMID: 40195349 PMCID: PMC11976975 DOI: 10.1038/s41598-025-90735-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 02/14/2025] [Indexed: 04/09/2025] Open
Abstract
Myocardial ischemic preconditioning (IPC) enhances myocardial resilience to ischemic injury. Myocardial stunning is a transient, reversible dysfunction, while necrosis involves irreversible cell death. The relationship between IPC, stunning, and necrosis is not well understood, requiring further molecular investigation. This study aimed to investigate the proteomic changes associated with IPC, focusing on its relationship with myocardial stunning and necrosis. A novel 13.5-minute ischemia-reperfusion (I/R) rat model was specifically chosen to induce myocardial stunning, providing a unique approach to assess IPC effects in this context. Rats underwent either IPC with two 5-minute ischemia/reperfusion cycles followed by a 13.5-minute ischemic period or the procedure without IPC (no ischemic preconditioning, NIPC). Myocardial samples were collected at early (T1) and 4-hour post-reperfusion (T2) time points for proteomic analysis. Protein levels were quantified by differential labeling using TMTpro reagents, and subsequent liquid chromatography-mass spectrometry. IPC induced upregulation of proteins involved in endocytosis and Fc gamma R-mediated phagocytosis pathways at T1, while downregulating proteins related to tissue remodeling, immune response, and coagulation at T2. Conversely, NIPC exhibited upregulation of proteins associated with tissue damage and inflammation. IPC rats demonstrated enhanced leukocyte migration, complement activation, and immune response between T1 and T2. Consistent proteomic changes were observed between T1 and T2 in IPC vs. NIPC groups, and common alterations between IPC T2 vs. T1 and NIPC T2 vs. T1 comparisons underline shared pathways in cardiac complement and coagulation cascades. Our study reveals distinct proteomic changes induced by IPC in the context of myocardial stunning and necrosis. IPC activates early protective pathways, attenuates tissue damage and inflammation, and preserves myocardial function. These findings underscore IPC's reparative potential and identify myocardial stunning as an important, transient adaptation, which may have implications for supportive clinical management in I/R.
Collapse
Affiliation(s)
- Yalda Kakaei
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Shafaat Hussain
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Ahmed Elmahdy
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Evelin Berger
- Proteomics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Aaron Shekka Espinosa
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Valentyna Sevastianova
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Zahra Sheybani
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Amin Al-Awar
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Mana Kalani
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Sandeep Jha
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital/S, Gothenburg, Sweden
| | - Ermir Zulfaj
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
| | - Amirali Nejat
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
| | - Abhishek Jha
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Tetiana Pylova
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Maryna Krasnikova
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Erik Axel Andersson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Vagner Ramon Rodrigues Silva
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Elmir Omerovic
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital/S, Gothenburg, Sweden
| | - Björn Redfors
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital/S, Gothenburg, Sweden
| |
Collapse
|
4
|
Yan D, Zhan S, Guo C, Han J, Zhan L, Zhou Q, Bing D, Wang X. The role of myocardial regeneration, cardiomyocyte apoptosis in acute myocardial infarction: A review of current research trends and challenges. J Cardiol 2025; 85:283-292. [PMID: 39393490 DOI: 10.1016/j.jjcc.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024]
Abstract
PURPOSE This paper aims to review the research progress in repairing injury caused by acute myocardial infarction, focusing on myocardial regeneration, cardiomyocyte apoptosis, and fibrosis. The goal is to investigate the current research trends and challenges in the field of myocardial injury repair. METHODS The review delves into the latest research on myocardial regeneration, cardiomyocyte apoptosis, and fibrosis following acute myocardial infarction. It highlights stem cell transplantation and gene therapy as key areas of current research focus, while emphasizing the significance of cardiomyocyte apoptosis and fibrosis in the myocardial injury repair process. Additionally, the review addresses the challenges and unresolved issues that require further investigation in the field of myocardial injury repair. SUMMARY Acute myocardial infarction is a prevalent cardiovascular condition that results in myocardial damage necessitating repair. Myocardial regeneration plays a crucial role in repairing myocardial injury, with current research focusing on stem cell transplantation and gene therapy. Cardiomyocyte apoptosis and fibrosis are key factors in the repair process, significantly impacting the restoration of myocardial structure and function. Nonetheless, there remain numerous challenges and unresolved issues that warrant further investigation in the realm of myocardial injury repair.
Collapse
Affiliation(s)
- Dan Yan
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China; Wuhan Asia Heart Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, China; Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China.
| | - Shifang Zhan
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Chenyu Guo
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Jiawen Han
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Lin Zhan
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Qianyi Zhou
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Dan Bing
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoyan Wang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou, China.
| |
Collapse
|
5
|
Zhang M, Wei J, Sun Y, He C, Ma S, Pan X, Zhu X. The efferocytosis process in aging: Supporting evidence, mechanisms, and therapeutic prospects for age-related diseases. J Adv Res 2025; 69:31-49. [PMID: 38499245 PMCID: PMC11954809 DOI: 10.1016/j.jare.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Aging is characterized by an ongoing struggle between the buildup of damage caused by a combination of external and internal factors. Aging has different effects on phagocytes, including impaired efferocytosis. A deficiency in efferocytosis can cause chronic inflammation, aging, and several other clinical disorders. AIM OF REVIEW Our review underscores the possible feasibility and extensive scope of employing dual targets in various age-related diseases to reduce the occurrence and progression of age-related diseases, ultimately fostering healthy aging and increasing lifespan. Key scientific concepts of review Hence, the concurrent implementation of strategies aimed at augmenting efferocytic mechanisms and anti-aging treatments has the potential to serve as a potent intervention for extending the duration of a healthy lifespan. In this review, we comprehensively discuss the concept and physiological effects of efferocytosis. Subsequently, we investigated the association between efferocytosis and the hallmarks of aging. Finally, we discuss growing evidence regarding therapeutic interventions for age-related disorders, focusing on the physiological processes of aging and efferocytosis.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jin Wei
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Yu Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Chang He
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Shiyin Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
6
|
Fonseka O, Gare SR, Chen X, Zhang J, Alatawi NH, Ross C, Liu W. Molecular Mechanisms Underlying Heart Failure and Their Therapeutic Potential. Cells 2025; 14:324. [PMID: 40072053 PMCID: PMC11899429 DOI: 10.3390/cells14050324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/15/2025] Open
Abstract
Heart failure (HF) is a prominent fatal cardiovascular disorder afflicting 3.4% of the adult population despite the advancement of treatment options. Therefore, a better understanding of the pathogenesis of HF is essential for exploring novel therapeutic strategies. Hypertrophy and fibrosis are significant characteristics of pathological cardiac remodeling, contributing to HF. The mechanisms involved in the development of cardiac remodeling and consequent HF are multifactorial, and in this review, the key underlying mechanisms are discussed. These have been divided into the following categories thusly: (i) mitochondrial dysfunction, including defective dynamics, energy production, and oxidative stress; (ii) cardiac lipotoxicity; (iii) maladaptive endoplasmic reticulum (ER) stress; (iv) impaired autophagy; (v) cardiac inflammatory responses; (vi) programmed cell death, including apoptosis, pyroptosis, and ferroptosis; (vii) endothelial dysfunction; and (viii) defective cardiac contractility. Preclinical data suggest that there is merit in targeting the identified pathways; however, their clinical implications and outcomes regarding treating HF need further investigation in the future. Herein, we introduce the molecular mechanisms pivotal in the onset and progression of HF, as well as compounds targeting the related mechanisms and their therapeutic potential in preventing or rescuing HF. This, therefore, offers an avenue for the design and discovery of novel therapies for the treatment of HF.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Liu
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (O.F.); (S.R.G.); (X.C.); (J.Z.); (N.H.A.)
| |
Collapse
|
7
|
Astiawati T, Rohman MS, Wihastuti T, Sujuti H, Endharti AT, Sargowo D, Oceandy D, Lestari B, Triastuti E, Nugraha RA. Efficacy of Colchicine in Reducing NT-proBNP, Caspase-1, TGF-β, and Galectin-3 Expression and Improving Echocardiography Parameters in Acute Myocardial Infarction: A Multi-Center, Randomized, Placebo-Controlled, Double-Blinded Clinical Trial. J Clin Med 2025; 14:1347. [PMID: 40004876 PMCID: PMC11856086 DOI: 10.3390/jcm14041347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/07/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Caspase-1 (reflects NOD-like receptor protein 3 inflammasome activity), transforming growth factor-β (TGF-β), and Galectin-3 play significant roles in post-AMI fibrosis and inflammation. Recently, colchicine was shown to dampen inflammation after AMI; however, its direct benefit remains controversial. Objectives: This study aimed to analyze the benefit of colchicine in reducing NT-proBNP, Caspase-1, TGF-β,and Galectin-3 expression and improving systolic-diastolic echocardiography parameters among AMI patients. Methods: A double-blinded, placebo-controlled, randomized, multicenter clinical trial was conducted at three hospitals in East Java, Indonesia: Dr. Saiful Anwar Hospital Malang, Dr. Soebandi Hospital Jember, and Dr. Iskak Hospital Tulungagung, between 1 June and 31 December 2023. A total of 161 eligible AMI subjects were randomly allocated 1:1 to colchicine (0.5 mg daily) or standard treatment for 30 days. Caspase-1, TGF-β, and Galectin-3 were tested on day 1 and day 5 by ELISA, while NT-proBNP was tested on days 5 and 30. Transthoracic echocardiography was also performed on day 5 and day 30. Results: By day 30, no significant improvements in systolic-diastolic echocardiography parameters had been shown in the colchicine group. However, colchicine reduced the level of NT-proBNP on day 30 more than placebo (ΔNT-proBNP: -73.74 ± 87.53 vs. -75.75 ± 12.44 pg/mL; p < 0.001). Moreover, colchicine lowered the level of Caspase-1 expression on day 5 and the levels of TGF-β and Galectin-3 expression on day 1. Conclusions: Colchicine can reduce NT-proBNP, Caspase-1, TGF-β, and Galectin-3 expression significantly among AMI patients. Colchicine administration was capable of reducing post-AMI inflammation, ventricular dysfunction, and heart failure but did not improve systolic-diastolic echocardiography parameters (ClinicalTrials.gov identifier: NCT06426537).
Collapse
Affiliation(s)
- Tri Astiawati
- Doctoral Program of Medical Science, Brawijaya University, Malang 65145, Indonesia
- Department of Cardiology and Vascular Medicine, Dr. Iskak General Hospital, Tulungagung 66223, Indonesia
| | - Mohammad Saifur Rohman
- Department of Cardiology and Cardiovascular Medicine, Faculty of Medicine, Brawijaya University, Dr. Saiful Anwar General Hospital, Malang 65145, Indonesia;
- Cardiovascular Research Centre, Universitas Brawijaya, Malang 65145, Indonesia
| | - Titin Wihastuti
- Department of Nursing Science, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Hidayat Sujuti
- Department of Biochemistry, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Agustina Tri Endharti
- Department of Parasitology, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Djanggan Sargowo
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University, Dr. Saiful Anwar General Hospital, Malang 65145, Indonesia;
| | - Delvac Oceandy
- Division of Cardiovascular Science, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK;
| | - Bayu Lestari
- Department of Pharmacology, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Efta Triastuti
- Department of Pharmacy, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Ricardo Adrian Nugraha
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo General Hospital, Surabaya 60286, Indonesia;
| |
Collapse
|
8
|
Hu Y, Zhang W, Ali SR, Takeda K, Vahl TP, Zhu D, Hong Y, Cheng K. Extracellular vesicle therapeutics for cardiac repair. J Mol Cell Cardiol 2025; 199:12-32. [PMID: 39603560 PMCID: PMC11788051 DOI: 10.1016/j.yjmcc.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/30/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Extracellular vesicles (EVs) are cell-secreted heterogeneous vesicles that play crucial roles in intercellular communication and disease pathogenesis. Due to their non-tumorigenicity, low immunogenicity, and therapeutic potential, EVs are increasingly used in cardiac repair as cell-free therapy. There exist multiple steps for the design of EV therapies, and each step offers many choices to tune EV properties. Factors such as EV source, cargo, loading methods, routes of administration, surface modification, and biomaterials are comprehensively considered to achieve specific goals. PubMed and Google Scholar were searched in this review, 89 articles related to EV-based cardiac therapy over the past five years (2019 Jan - 2023 Dec) were included, and their key steps in designing EV therapies were counted and analyzed. We aim to provide a comprehensive overview that can serve as a reference guide for researchers to design EV-based cardiac therapies.
Collapse
Affiliation(s)
- Yilan Hu
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Weihang Zhang
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Shah Rukh Ali
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Koji Takeda
- Division of Cardiac Surgery, Department of Surgery, Columbia University, New York, NY 10032, USA
| | - Torsten Peter Vahl
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Donghui Zhu
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Ke Cheng
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
9
|
Zhang Z, Du H, Gao W, Zhang D. Engineered macrophages: an "Intelligent Repair" cellular machine for heart injury. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:25. [PMID: 39592532 PMCID: PMC11599506 DOI: 10.1186/s13619-024-00209-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/22/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024]
Abstract
Macrophages are crucial in the heart's development, function, and injury. As part of the innate immune system, they act as the first line of defense during cardiac injury and repair. After events such as myocardial infarction or myocarditis, numerous macrophages are recruited to the affected areas of the heart to clear dead cells and facilitate tissue repair. This review summarizes the roles of resident and recruited macrophages in developing cardiovascular diseases. We also describe how macrophage phenotypes dynamically change within the cardiovascular disease microenvironment, exhibiting distinct pro-inflammatory and anti-inflammatory functions. Recent studies reveal the values of targeting macrophages in cardiovascular diseases treatment and the novel bioengineering technologies facilitate engineered macrophages as a promising therapeutic strategy. Engineered macrophages have strong natural tropism and infiltration for cardiovascular diseases aiming to reduce inflammatory response, inhibit excessive fibrosis, restore heart function and promote heart regeneration. We also discuss recent studies highlighting therapeutic strategies and new approaches targeting engineered macrophages, which can aid in heart injury recovery.
Collapse
Affiliation(s)
- Zhuo Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, 430062, China
- Stem Cells and Tissue Engineering Manufacture Center, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Hetian Du
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, 430062, China
- Stem Cells and Tissue Engineering Manufacture Center, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Weijie Gao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, 430062, China.
- Stem Cells and Tissue Engineering Manufacture Center, School of Life Sciences, Hubei University, Wuhan, 430062, China.
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, 430062, China.
- Stem Cells and Tissue Engineering Manufacture Center, School of Life Sciences, Hubei University, Wuhan, 430062, China.
| |
Collapse
|
10
|
Martins RA, Costa FR, Pires L, Santos M, Santos GS, Lana JV, Costa BR, Santos N, de Macedo AP, Kruel A, Lana JF. Regenerative Inflammation: The Mechanism Explained from the Perspective of Buffy-Coat Protagonism and Macrophage Polarization. Int J Mol Sci 2024; 25:11329. [PMID: 39457111 PMCID: PMC11508762 DOI: 10.3390/ijms252011329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
The buffy-coat, a layer of leukocytes and platelets obtained from peripheral blood centrifugation, plays a crucial role in tissue regeneration and the modulation of inflammatory responses. This article explores the mechanisms of regenerative inflammation, highlighting the critical role of the buffy-coat in influencing macrophage polarization and its therapeutic potential. Macrophage polarization into M1 and M2 subtypes is pivotal in balancing inflammation and tissue repair, with M1 macrophages driving pro-inflammatory responses and M2 macrophages promoting tissue healing and regeneration. The buffy-coat's rich composition of progenitor cells, cytokines, and growth factors-such as interleukin-10, transforming growth factor-β, and monocyte colony-stimulating factor-supports the transition from M1 to M2 macrophages, enhancing tissue repair and the resolution of inflammation. This dynamic interaction between buffy-coat components and macrophages opens new avenues for therapeutic strategies aimed at improving tissue regeneration and managing inflammatory conditions, particularly in musculoskeletal diseases such as osteoarthritis. Furthermore, the use of buffy-coat-derived therapies in conjunction with other regenerative modalities, such as platelet-rich plasma, holds promise for more effective clinical outcomes.
Collapse
Affiliation(s)
| | - Fábio Ramos Costa
- Department of Orthopedics, FC Sports Traumatology, Salvador 40296-210, Brazil;
| | - Luyddy Pires
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, Brazil; (L.P.); (N.S.); (A.P.d.M.); (J.F.L.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, Brazil;
| | - Márcia Santos
- Nutritional Sciences, Metropolitan Union of Education and Culture, Salvador 42700-000, Brazil;
| | - Gabriel Silva Santos
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, Brazil; (L.P.); (N.S.); (A.P.d.M.); (J.F.L.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, Brazil;
| | - João Vitor Lana
- Medical School, Max Planck University Center (UniMAX), Indaiatuba 13343-060, Brazil;
| | | | - Napoliane Santos
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, Brazil; (L.P.); (N.S.); (A.P.d.M.); (J.F.L.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, Brazil;
| | - Alex Pontes de Macedo
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, Brazil; (L.P.); (N.S.); (A.P.d.M.); (J.F.L.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, Brazil;
| | - André Kruel
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, Brazil;
| | - José Fábio Lana
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, Brazil; (L.P.); (N.S.); (A.P.d.M.); (J.F.L.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, Brazil;
- Medical School, Max Planck University Center (UniMAX), Indaiatuba 13343-060, Brazil;
- Clinical Research, Anna Vitória Lana Institute (IAVL), Indaiatuba 13334-170, Brazil
- Medical School, Jaguariúna University Center (UniFAJ), Jaguariúna 13911-094, Brazil
| |
Collapse
|
11
|
Feng Y, Wang Y, Li L, Yang Y, Tan X, Chen T. Exosomes Induce Crosstalk Between Multiple Types of Cells and Cardiac Fibroblasts: Therapeutic Potential for Remodeling After Myocardial Infarction. Int J Nanomedicine 2024; 19:10605-10621. [PMID: 39445157 PMCID: PMC11498042 DOI: 10.2147/ijn.s476995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024] Open
Abstract
Recanalization therapy can significantly improve the prognosis of patients with acute myocardial infarction (AMI). However, infarction or reperfusion-induced cardiomyocyte death, immune cell infiltration, fibroblast proliferation, and scarring formation lead to cardiac remodeling and gradually progress to heart failure or arrhythmia, resulting in a high mortality rate. Due to the inability of cardiomyocytes to regenerate, the healing of infarcted myocardium mainly relies on the formation of scars. Cardiac fibroblasts, as the main effector cells involved in repair and scar formation, play a crucial role in maintaining the structural integrity of the heart after MI. Recent studies have revealed that exosome-mediated intercellular communication plays a huge role in myocardial repair and signaling transduction after myocardial infarction (MI). Exosomes can regulate the biological behavior of fibroblasts by activating or inhibiting the intracellular signaling pathways through their contents, which are derived from cardiomyocytes, immune cells, endothelial cells, mesenchymal cells, and others. Understanding the interactions between fibroblasts and other cell types during cardiac remodeling will be the key to breakthrough therapies. This review examines the role of exosomes from different sources in the repair process after MI injury, especially the impacts on fibroblasts during myocardial remodeling, and explores the use of exosomes in the treatment of myocardial remodeling after MI.
Collapse
Affiliation(s)
- Yijuan Feng
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yan Wang
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Li Li
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Xiaoqiu Tan
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Tangting Chen
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| |
Collapse
|
12
|
Liao L, Wang T, Zhang L, Wei Y, Fan X. Protective Mechanisms of SGLTi in Ischemic Heart Disease. J Cardiovasc Transl Res 2024; 17:1018-1035. [PMID: 38767796 DOI: 10.1007/s12265-024-10513-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/11/2024] [Indexed: 05/22/2024]
Abstract
Ischemic heart disease (IHD) is a common clinical cardiovascular disease with high morbidity and mortality. Sodium glucose cotransporter protein inhibitor (SGLTi) is a novel hypoglycemic drug. To date, both clinical trials and animal experiments have shown that SGLTi play a protective role in IHD, including myocardial infarction (MI) and ischemia/reperfusion (I/R). The protective effects may be involved in mechanisms of energy metabolic conversion, anti-inflammation, anti-fibrosis, ionic homeostasis improvement, immune cell development, angiogenesis and functional regulation, gut microbiota regulation, and epicardial lipids. Thus, this review summarizes the above mechanisms and aims to provide theoretical evidence for therapeutic strategies for IHD.
Collapse
Affiliation(s)
- Lei Liao
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Tong Wang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Lu Zhang
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yan Wei
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Xinrong Fan
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
13
|
Xu Y, Wang Y, Ning K, Bao Y. Unraveling the Mechanisms of S100A8/A9 in Myocardial Injury and Dysfunction. Curr Issues Mol Biol 2024; 46:9707-9720. [PMID: 39329929 PMCID: PMC11429546 DOI: 10.3390/cimb46090577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 08/22/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
S100A8 and S100A9, which are prominent members of the calcium-binding protein S100 family and recognized as calprotectin, form a robust heterodimer known as S100A8/A9, crucial for the manifestation of their diverse biological effects. Currently, there is a consensus that S100A8/A9 holds promise as a biomarker for cardiovascular diseases (CVDs), exerting an influence on cardiomyocytes or the cardiovascular system through multifaceted mechanisms that contribute to myocardial injury or dysfunction. In particular, the dualistic nature of S100A8/A9, which functions as both an inflammatory mediator and an anti-inflammatory agent, has garnered significantly increasing attention. This comprehensive review explores the intricate mechanisms through which S100A8/A9 operates in cardiovascular diseases, encompassing its bidirectional regulatory role in inflammation, the initiation of mitochondrial dysfunction, the dual modulation of myocardial fibrosis progression, and apoptosis and autophagy. The objective is to provide new information on and strategies for the clinical diagnosis and treatment of cardiovascular diseases in the future.
Collapse
Affiliation(s)
| | | | | | - Yimin Bao
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Shanghai 201203, China; (Y.X.); (Y.W.); (K.N.)
| |
Collapse
|
14
|
Chen R, Bao Q, Ma X. Association of IL13 polymorphisms with susceptibility to myocardial infarction: A case-control study in Chinese population. PLoS One 2024; 19:e0308081. [PMID: 39088580 PMCID: PMC11293651 DOI: 10.1371/journal.pone.0308081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/16/2024] [Indexed: 08/03/2024] Open
Abstract
BACKGROUND Inflammatory cytokines play a major role in the pathogenesis of myocardial infarction (MI). Although information on the importance of interleukin 13 (IL13) in human MI is limited, it has been well documented in the mouse model. Genetic variation in the IL13 gene has been associated with the structure and expression of the IL13. In the present study, we hypothesized that IL13 common genetic variants would be associated with a predisposition to the development of MI. MATERIALS AND METHODS The present study enrolled 305 MI patients and 310 matched healthy controls. Common genetic polymorphisms in the IL13 gene (rs20541, rs1881457, and rs1800925) were genotyped using the TaqMan SNP genotyping method. Plasma levels of IL13 were measured using an enzyme-linked immunosorbent assay (ELISA). RESULTS In MI patients, minor alleles of the IL13 rs1881457 and rs1800925 polymorphisms were less common than in healthy controls [rs1881457: AC (P = 0.004, OR = 0.61), C (P = 0.001, OR = 0.66); rs1800925: CT (P = 0.006, OR = 0.59)]. Further haplotype analysis of three studied SNPs revealed a significant association with predisposition to MI. Interestingly, IL13 rs1881457 and rs1800925 were linked to plasma levels of IL13: the reference genotype had higher levels, heterozygotes were intermediate, and the alternate genotype had the lowest levels. CONCLUSIONS In the Chinese population, IL13 (rs1881457 and rs180092) variants are associated with different plasma IL13 levels and offer protection against MI development. However, additional research is required to validate our findings in different populations, including descent samples.
Collapse
Affiliation(s)
- Rong Chen
- Department of Cardiology, Qinghai Province Cardiovascular and Cerebrovascular Disease Specialist Hospital, Xining, Qinghai, China
| | - Qiaoling Bao
- Department of Coronary Heart Disease, Qinghai Province Cardiovascular and Cerebrovascular Disease Specialist Hospital, Xining, Qinghai, China
| | - Xiaofeng Ma
- Department of Cardiology, Qinghai Province Cardiovascular and Cerebrovascular Disease Specialist Hospital, Xining, Qinghai, China
| |
Collapse
|
15
|
Zuo B, Fan X, Xu D, Zhao L, Zhang B, Li X. Deciphering the mitochondria-inflammation axis: Insights and therapeutic strategies for heart failure. Int Immunopharmacol 2024; 139:112697. [PMID: 39024750 DOI: 10.1016/j.intimp.2024.112697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/10/2024] [Accepted: 07/13/2024] [Indexed: 07/20/2024]
Abstract
Heart failure (HF) is a clinical syndrome resulting from left ventricular systolic and diastolic dysfunction, leading to significant morbidity and mortality worldwide. Despite improvements in medical treatment, the prognosis of HF patients remains unsatisfactory, with high rehospitalization rates and substantial economic burdens. The heart, a high-energy-consuming organ, relies heavily on ATP production through oxidative phosphorylation in mitochondria. Mitochondrial dysfunction, characterized by impaired energy production, oxidative stress, and disrupted calcium homeostasis, plays a crucial role in HF pathogenesis. Additionally, inflammation contributes significantly to HF progression, with elevated levels of circulating inflammatory cytokines observed in patients. The interplay between mitochondrial dysfunction and inflammation involves shared risk factors, signaling pathways, and potential therapeutic targets. This review comprehensively explores the mechanisms linking mitochondrial dysfunction and inflammation in HF, including the roles of mitochondrial reactive oxygen species (ROS), calcium dysregulation, and mitochondrial DNA (mtDNA) release in triggering inflammatory responses. Understanding these complex interactions offers insights into novel therapeutic approaches for improving mitochondrial function and relieving oxidative stress and inflammation. Targeted interventions that address the mitochondria-inflammation axis hold promise for enhancing cardiac function and outcomes in HF patients.
Collapse
Affiliation(s)
- Baile Zuo
- Molecular Immunology and Immunotherapy Laboratory, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiu Fan
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Dawei Xu
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Liping Zhao
- Department of Pathology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Bi Zhang
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China.
| | - Xiaoyan Li
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China; Department of Clinical Laboratory, Heping Branch, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China.
| |
Collapse
|
16
|
Barbu E, Mihaila A, Filippi A, Stoenescu A, Ciortan L, Butoi E, Beiu C, Popescu MN, Balanescu S. Stress, Hyperglycemia, and Insulin Resistance Correlate With Neutrophil Activity and Impact Acute Myocardial Infarction Outcomes. Cureus 2024; 16:e63731. [PMID: 39100008 PMCID: PMC11295428 DOI: 10.7759/cureus.63731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/03/2024] [Indexed: 08/06/2024] Open
Abstract
Introduction Acute insulin resistance (IR) and hyperglycemia are frequently observed during acute myocardial infarction (AMI), significantly influencing both immediate and long-term patient outcomes, irrespective of diabetic status. Neutrophilia and increased neutrophil activity, which are common in these scenarios, have been associated with poorer prognoses, as demonstrated in our recent findings. While it is well established that neutrophils and stress-induced hyperglycemia exacerbate inflammation and hinder recovery, the complex interplay between these factors and their combined impact on AMI prognosis remains inadequately understood. This study aims to investigate the effects of stress hyperglycemia and IR on AMI patients at the onset of the event and to elucidate the relationship between these metabolic disturbances and inflammatory markers, particularly neutrophils. Methods We conducted a longitudinal prospective study on 219 AMI patients at Elias Emergency Hospital in Bucharest, Romania, from April 2021 to September 2022. Patients were included within 24 hours of AMI with ST-segment elevation and excluded if they had acute infections or chronic inflammatory diseases. Blood samples were collected to study inflammatory biomarkers, including neutrophil extracellular traps (NETs), S100A8/A9, interleukin (IL)-1β, IL-18, and IL-6. Diabetic and pre-diabetic statuses were defined using glycated hemoglobin (HbA1c) and medical history (ADA 2019 criteria). To assess glycemic parameters, we employed the glycemia ratio (GR) and the homeostatic model assessment of insulin resistance (HOMA-IR) index, enabling a precise evaluation of stress hyperglycemia, acute IR, and their prognostic implications. Patients were stratified into groups based on GR calculations, categorized as under-average glycemia, normal glycemia, and stress hyperglycemia. Results The majority of patients in the stress hyperglycemia group exhibited an unfavorable prognosis. This group also demonstrated significantly elevated neutrophil counts and neutrophil-to-lymphocyte ratios (NLR). The GR was significantly and positively correlated with inflammation markers, including neutrophil count (Pearson's R = 0.181, P = 0.008) and NLR (Pearson's R = 0.318, P < 0.001), but showed no significant correlation with other evaluated inflammatory markers. Conclusions Our findings suggest that poor outcomes in AMI patients may be associated with stress hyperglycemia, as indicated by GR. AcuteIR, quantified by GR and HOMA-IR, exhibits a strong correlation with neutrophil count and NLR within the first 24 hours of AMI onset. However, no significant correlation was observed with other inflammatory markers, such as IL-1β, IL-18, and IL-6, underscoring the specific interplay between IR and neutrophil activity in this setting.
Collapse
Affiliation(s)
- Elena Barbu
- Department of Cardiology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Andreea Mihaila
- Department of Inflammation, Institute of Cellular Biology and Pathology Nicolae Simionescu, Bucharest, ROU
| | - Alexandru Filippi
- Department of Biochemistry and Biophysics, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Andra Stoenescu
- Department of Cardiology, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Letitia Ciortan
- Department of Inflammation, Institute of Cellular Biology and Pathology Nicolae Simionescu, Bucharest, ROU
| | - Elena Butoi
- Department of Inflammation, Institute of Cellular Biology and Pathology Nicolae Simionescu, Bucharest, ROU
| | - Cristina Beiu
- Department of Oncologic Dermatology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Marius N Popescu
- Department of Physical Medicine and Rehabilitation, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Serban Balanescu
- Department of Cardiology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| |
Collapse
|
17
|
Wang X, Yang G, Li J, Meng C, Xue Z. Dynamic molecular signatures of acute myocardial infarction based on transcriptomics and metabolomics. Sci Rep 2024; 14:10175. [PMID: 38702356 PMCID: PMC11068872 DOI: 10.1038/s41598-024-60945-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/29/2024] [Indexed: 05/06/2024] Open
Abstract
Acute myocardial infarction (AMI) commonly precedes ventricular remodeling, heart failure. Few dynamic molecular signatures have gained widespread acceptance in mainstream clinical testing despite the discovery of many potential candidates. These unmet needs with respect to biomarker and drug discovery of AMI necessitate a prioritization. We enrolled patients with AMI aged between 30 and 70. RNA-seq analysis was performed on the peripheral blood mononuclear cells collected from the patients at three time points: 1 day, 7 days, and 3 months after AMI. PLC/LC-MS analysis was conducted on the peripheral blood plasma collected from these patients at the same three time points. Differential genes and metabolites between groups were screened by bio-informatics methods to understand the dynamic changes of AMI in different periods. We obtained 15 transcriptional and 95 metabolite expression profiles at three time points after AMI through high-throughput sequencing. AMI-1d: enrichment analysis revealed the biological features of 1 day after AMI primarily included acute inflammatory response, elevated glycerophospholipid metabolism, and decreased protein synthesis capacity. Phosphatidylcholine (PC) and phosphatidylethanolamine (PE) might stand promising biomarkers to differentiate post-AMI stage. Anti-inflammatory therapy during the acute phase is an important direction for preventing related pathology. AMI-7d: the biological features of this stage primarily involved the initiation of cardiac fibrosis response and activation of platelet adhesion pathways. Accompanied by upregulated TGF-beta signaling pathway and ECM receptor interaction, GP5 help assess platelet activation, a potential therapeutic target to improve haemostasis. AMI-3m: the biological features of 3 months after AMI primarily showed a vascular regeneration response with VEGF signaling pathway, NOS3 and SHC2 widely activated, which holds promise for providing new therapeutic approaches for AMI. Our analysis highlights transcriptional and metabolomics signatures at different time points after MI, which deepens our understanding of the dynamic biological responses and associated molecular mechanisms that occur during cardiac repair.
Collapse
Affiliation(s)
- Xuejiao Wang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Guang Yang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Jun Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5 Beixiange, Xicheng District, Beijing, 100053, China.
| | - Chao Meng
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Zengming Xue
- Department of Cardiology, Langfang People's Hospital, Hebei Medical University, No. 37, Xinhua Road, Langfang, 065000, China.
| |
Collapse
|
18
|
Luan Y, Luan Y, Jiao Y, Liu H, Huang Z, Feng Q, Pei J, Yang Y, Ren K. Broadening Horizons: Exploring mtDAMPs as a Mechanism and Potential Intervention Target in Cardiovascular Diseases. Aging Dis 2023; 15:2395-2416. [PMID: 38270118 PMCID: PMC11567272 DOI: 10.14336/ad.2023.1130] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/30/2023] [Indexed: 01/26/2024] Open
Abstract
Cardiovascular diseases (CVDs) have been recognized as the leading cause of premature mortality and morbidity worldwide despite significant advances in therapeutics. Inflammation is a key factor in CVD progression. Once stress stimulates cells, they release cellular compartments known as damage-associated molecular patterns (DAMPs). Mitochondria can release mitochondrial DAMPs (mtDAMPs) to initiate an immune response when stimulated with cellular stress. Investigating the molecular mechanisms underlying the DAMPs that regulate CVD progression is crucial for improving CVDs. Herein, we discuss the composition and mechanism of DAMPs, the significance of mtDAMPs in cellular inflammation, the presence of mtDAMPs in different types of cells, and the main signaling pathways associated with mtDAMPs. Based on this, we determined the role of DAMPs in CVDs and the effects of mtDAMP intervention on CVD progression. By offering a fresh perspective and comprehensive insights into the molecular mechanisms of DAMPs, this review seeks to provide important theoretical foundations for developing drugs targeting CVDs.
Collapse
Affiliation(s)
- Yi Luan
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Ying Luan
- State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, China.
| | - Yuxue Jiao
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Hui Liu
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China.
| | - Zhen Huang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China.
| | - Qi Feng
- Department ofIntegrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Jinyan Pei
- Quality Management Department, The Third People’s Hospital of Henan Provine, Zhengzhou, China.
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
19
|
Wu L, Yin W, Wen J, Wang S, Li H, Wang X, Zhang W, Duan S, Zhu Q, Gao E, Wu S, Zhan B, Zhou R, Yang X. Excretory/secretory products from Trichinella spiralis adult worms ameliorate myocardial infarction by inducing M2 macrophage polarization in a mouse model. Parasit Vectors 2023; 16:362. [PMID: 37845695 PMCID: PMC10577921 DOI: 10.1186/s13071-023-05930-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/14/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Ischemia-induced inflammatory response is the main pathological mechanism of myocardial infarction (MI)-caused heart tissue injury. It has been known that helminths and worm-derived proteins are capable of modulating host immune response to suppress excessive inflammation as a survival strategy. Excretory/secretory products from Trichinella spiralis adult worms (Ts-AES) have been shown to ameliorate inflammation-related diseases. In this study, Ts-AES were used to treat mice with MI to determine its therapeutic effect on reducing MI-induced heart inflammation and the immunological mechanism involved in the treatment. METHODS The MI model was established by the ligation of the left anterior descending coronary artery, followed by the treatment of Ts-AES by intraperitoneal injection. The therapeutic effect of Ts-AES on MI was evaluated by measuring the heart/body weight ratio, cardiac systolic and diastolic functions, histopathological change in affected heart tissue and observing the 28-day survival rate. The effect of Ts-AES on mouse macrophage polarization was determined by stimulating mouse bone marrow macrophages in vitro with Ts-AES, and the macrophage phenotype was determined by flow cytometry. The protective effect of Ts-AES-regulated macrophage polarization on hypoxic cardiomyocytes was determined by in vitro co-culturing Ts-AES-induced mouse bone marrow macrophages with hypoxic cardiomyocytes and cardiomyocyte apoptosis determined by flow cytometry. RESULTS We observed that treatment with Ts-AES significantly improved cardiac function and ventricular remodeling, reduced pathological damage and mortality in mice with MI, associated with decreased pro-inflammatory cytokine levels, increased regulatory cytokine expression and promoted macrophage polarization from M1 to M2 type in MI mice. Ts-AES-induced M2 macrophage polarization also reduced apoptosis of hypoxic cardiomyocytes in vitro. CONCLUSIONS Our results demonstrate that Ts-AES ameliorates MI in mice by promoting the polarization of macrophages toward the M2 type. Ts-AES is a potential pharmaceutical agent for the treatment of MI and other inflammation-related diseases.
Collapse
Affiliation(s)
- Lingqin Wu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
- Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, China
| | - Wenhui Yin
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Jutai Wen
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Shuying Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Huihui Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- Basic Medical College of Bengbu Medical College, Bengbu, 233000, China
| | - Xiaoli Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- Basic Medical College of Bengbu Medical College, Bengbu, 233000, China
| | - Weixiao Zhang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Shuyao Duan
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Qiuyu Zhu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Erhe Gao
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Shili Wu
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Rui Zhou
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China.
| | - Xiaodi Yang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China.
- Basic Medical College of Bengbu Medical College, Bengbu, 233000, China.
| |
Collapse
|